Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.943
Filtrer
1.
Sci Rep ; 14(1): 13367, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862693

RÉSUMÉ

Patients with distant metastasis of head and neck squamous cell carcinoma (HNSCC) often have a poor prognosis. However, early diagnosis of distant metastasis is challenging in clinical practice, and distant metastasis is often only detected in the late stages of tumor metastasis through imaging techniques. In this study, we utilized data from HNSCC patients collected from the TCGA database. Patients were divided into distant metastasis and nonmetastasis groups based on the tumor-node-metastasis (TNM) stage. We analyzed the differentially expressed genes between the two groups (DM/non-M DEGs) and their associated lncRNAs and generated a predictive model based on 23 lncRNAs that were significantly associated with the occurrence of distant metastasis in HNSCC patients. On this basis, we built a nomogram to predict the distant metastasis of HNSCC patients. Moreover, through WGCNA and Cytoscape software analysis of DM/non-M DEGs, we identified the gene most closely related to HNSCC distant metastasis: EIF5A. Our findings were validated using GEO data; EIF5A expression was significantly increased in the tumor tissues of HNSCC patients with distant metastasis. We then predicted miRNAs that can directly bind to EIF5A via the TargetScan and miRWalk websites, intersected them with differentially expressed miRNAs in the two groups from the TCGA cohort, and identified the only overlapping miRNA, miR-424; we predicted the direct binding site of EIF5A and miR-424 via the miRWalk website. Immunohistochemistry further revealed high expression of EIF5A in the primary tumor tissue of HNSCC patients with distant metastasis. These results provide a new perspective for the early diagnosis of distant metastasis in HNSCC patients and the study of the mechanisms underlying HNSCC distant metastasis.


Sujet(s)
, Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou , Métastase tumorale , Nomogrammes , Facteurs initiation chaîne peptidique , Protéines de liaison à l'ARN , Carcinome épidermoïde de la tête et du cou , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Analyse de profil d'expression de gènes , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , microARN/génétique , microARN/métabolisme , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/métabolisme , Pronostic , ARN long non codant/génétique , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie
2.
Nat Cell Biol ; 26(6): 892-902, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38741019

RÉSUMÉ

Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a CAG trinucleotide repeat in the Huntingtin (HTT) gene, encoding a homopolymeric polyglutamine (polyQ) tract. Although mutant HTT (mHTT) protein is known to aggregate, the links between aggregation and neurotoxicity remain unclear. Here we show that both translation and aggregation of wild-type HTT and mHTT are regulated by a stress-responsive upstream open reading frame and that polyQ expansions cause abortive translation termination and release of truncated, aggregation-prone mHTT fragments. Notably, we find that mHTT depletes translation elongation factor eIF5A in brains of symptomatic HD mice and cultured HD cells, leading to pervasive ribosome pausing and collisions. Loss of eIF5A disrupts homeostatic controls and impairs recovery from acute stress. Importantly, drugs that inhibit translation initiation reduce premature termination and mitigate this escalating cascade of ribotoxic stress and dysfunction in HD.


Sujet(s)
, Protéine huntingtine , Maladie de Huntington , Facteurs initiation chaîne peptidique , Peptides , Homéostasie protéique , Protéines de liaison à l'ARN , Ribosomes , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Animaux , Peptides/métabolisme , Peptides/génétique , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Humains , Ribosomes/métabolisme , Ribosomes/génétique , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Souris transgéniques , Modèles animaux de maladie humaine , Stress physiologique , Encéphale/métabolisme , Encéphale/anatomopathologie , Expansion de trinucléotide répété/génétique
3.
Oncogene ; 43(24): 1824-1835, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38654106

RÉSUMÉ

We have performed a functional in vivo mutagenesis screen to identify genes that, when altered, cooperate with a heterozygous Pten mutation to promote prostate tumour formation. Two genes, Bzw2 and Eif5a2, which have been implicated in the process of protein translation, were selected for further validation. Using prostate organoid models, we show that either Bzw2 downregulation or EIF5A2 overexpression leads to increased organoid size and in vivo prostate growth. We show that both genes impact the PI3K pathway and drive a sustained increase in phospho-AKT expression, with PTEN protein levels reduced in both models. Mechanistic studies reveal that EIF5A2 is directly implicated in PTEN protein translation. Analysis of patient datasets identified EIF5A2 amplifications in many types of human cancer, including the prostate. Human prostate cancer samples in two independent cohorts showed a correlation between increased levels of EIF5A2 and upregulation of a PI3K pathway gene signature. Consistent with this, organoids with high levels of EIF5A2 were sensitive to AKT inhibitors. Our study identified novel genes that promote prostate cancer formation through upregulation of the PI3K pathway, predicting a strategy to treat patients with genetic aberrations in these genes particularly relevant for EIF5A2 amplified tumours.


Sujet(s)
, Phosphohydrolase PTEN , Facteurs initiation chaîne peptidique , Phosphatidylinositol 3-kinases , Tumeurs de la prostate , Protéines de liaison à l'ARN , Transduction du signal , Mâle , Humains , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/métabolisme , Transduction du signal/génétique , Animaux , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Souris , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Régulation de l'expression des gènes tumoraux , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Lignée cellulaire tumorale
4.
Exp Mol Med ; 56(5): 1123-1136, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38689086

RÉSUMÉ

Tumor-associated macrophages (TAMs) are vital contributors to the growth, metastasis, and therapeutic resistance of various cancers, including hepatocellular carcinoma (HCC). However, the exact phenotype of TAMs and the mechanisms underlying their modulation for therapeutic purposes have not been determined. Here, we present compelling evidence that glutamine-derived aspartate in TAMs stimulates spermidine production through the polyamine synthesis pathway, thereby increasing the translation efficiency of HIF-1α via eIF5A hypusination. Consequently, augmented translation of HIF-1α drives TAMs to undergo an increase glycolysis and acquire a metabolic phenotype distinct from that of M2 macrophages. Finally, eIF5A levels in tumor stromal lesions were greater than those in nontumor stromal lesions. Additionally, a higher degree of tumor stromal eIF5A hypusination was significantly associated with a more advanced tumor stage. Taken together, these data highlight the potential of inhibiting hypusinated eIF5A by targeting glutamine metabolism in TAMs, thereby opening a promising avenue for the development of novel therapeutic approaches for HCC.


Sujet(s)
Acide aspartique , Carcinome hépatocellulaire , , Glutamine , Sous-unité alpha du facteur-1 induit par l'hypoxie , Tumeurs du foie , Facteurs initiation chaîne peptidique , Protéines de liaison à l'ARN , Macrophages associés aux tumeurs , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Glutamine/métabolisme , Acide aspartique/métabolisme , Acide aspartique/analogues et dérivés , Biosynthèse des protéines , Animaux , Lignée cellulaire tumorale , Souris , Glycolyse , Lysine/analogues et dérivés
5.
Breast Cancer Res ; 26(1): 70, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38654332

RÉSUMÉ

BACKGROUND: Basal-like breast cancer (BLBC) is the most aggressive subtype of breast cancer due to its aggressive characteristics and lack of effective therapeutics. However, the mechanism underlying its aggressiveness remains largely unclear. S-adenosylmethionine decarboxylase proenzyme (AMD1) overexpression occurs specifically in BLBC. Here, we explored the potential molecular mechanisms and functions of AMD1 promoting the aggressiveness of BLBC. METHODS: The potential effects of AMD1 on breast cancer cells were tested by western blotting, colony formation, cell proliferation assay, migration and invasion assay. The spermidine level was determined by high performance liquid chromatography. The methylation status of CpG sites within the AMD1 promoter was evaluated by bisulfite sequencing PCR. We elucidated the relationship between AMD1 and Sox10 by ChIP assays and quantitative real-time PCR. The effect of AMD1 expression on breast cancer cells was evaluated by in vitro and in vivo tumorigenesis model. RESULTS: In this study, we showed that AMD1 expression was remarkably elevated in BLBC. AMD1 copy number amplification, hypomethylation of AMD1 promoter and transcription activity of Sox10 contributed to the overexpression of AMD1 in BLBC. AMD1 overexpression enhanced spermidine production, which enhanced eIF5A hypusination, activating translation of TCF4 with multiple conserved Pro-Pro motifs. Our studies showed that AMD1-mediated metabolic system of polyamine in BLBC cells promoted tumor cell proliferation and tumor growth. Clinically, elevated expression of AMD1 was correlated with high grade, metastasis and poor survival, indicating poor prognosis of breast cancer patients. CONCLUSION: Our work reveals the critical association of AMD1-mediated spermidine-eIF5A hypusination-TCF4 axis with BLBC aggressiveness, indicating potential prognostic indicators and therapeutic targets for BLBC.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , , Régulation de l'expression des gènes tumoraux , Lysine/analogues et dérivés , Facteurs initiation chaîne peptidique , Protéines de liaison à l'ARN , Spermidine , Facteur-4 de transcription , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/mortalité , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/génétique , Souris , Animaux , Spermidine/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Facteur-4 de transcription/métabolisme , Facteur-4 de transcription/génétique , Lignée cellulaire tumorale , Régions promotrices (génétique) , Adenosylmethionine decarboxylase/métabolisme , Adenosylmethionine decarboxylase/génétique , Mouvement cellulaire/génétique , Méthylation de l'ADN , Pronostic , Facteurs de transcription SOX-E/métabolisme , Facteurs de transcription SOX-E/génétique
6.
Neurobiol Dis ; 195: 106488, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38565397

RÉSUMÉ

Given their highly polarized morphology and functional singularity, neurons require precise spatial and temporal control of protein synthesis. Alterations in protein translation have been implicated in the development and progression of a wide range of neurological and neurodegenerative disorders, including Huntington's disease (HD). In this study we examined the architecture of polysomes in their native brain context in striatal tissue from the zQ175 knock-in mouse model of HD. We performed 3D electron tomography of high-pressure frozen and freeze-substituted striatal tissue from HD models and corresponding controls at different ages. Electron tomography results revealed progressive remodelling towards a more compacted polysomal architecture in the mouse model, an effect that coincided with the emergence and progression of HD related symptoms. The aberrant polysomal architecture is compatible with ribosome stalling phenomena. In fact, we also detected in the zQ175 model an increase in the striatal expression of the stalling relief factor EIF5A2 and an increase in the accumulation of eIF5A1, eIF5A2 and hypusinated eIF5A1, the active form of eIF5A1. Polysomal sedimentation gradients showed differences in the relative accumulation of 40S ribosomal subunits and in polysomal distribution in striatal samples of the zQ175 model. These findings indicate that changes in the architecture of the protein synthesis machinery may underlie translational alterations associated with HD, opening new avenues for understanding the progression of the disease.


Sujet(s)
Modèles animaux de maladie humaine , Maladie de Huntington , Polyribosomes , Ribosomes , Animaux , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Maladie de Huntington/génétique , Souris , Polyribosomes/métabolisme , Ribosomes/métabolisme , Corps strié/métabolisme , Corps strié/anatomopathologie , Souris transgéniques , Évolution de la maladie , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/génétique
7.
Medicina (Kaunas) ; 60(4)2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38674185

RÉSUMÉ

Oxidative stress is implicated in the pathogenesis of various acute disorders including ischemia/reperfusion injury, ultraviolet/radiation burn, as well as chronic disorders such as dyslipidemia, atherosclerosis, diabetes mellitus, chronic renal disease, and inflammatory bowel disease (IBD). However, the precise mechanism involved remains to be clarified. We formerly identified a novel apoptosis-inducing humoral protein, in a hypoxia/reoxygenation-conditioned medium of cardiac myocytes, which proved to be 69th tyrosine-sulfated eukaryotic translation initiation factor 5A (eIF5A). We named this novel tyrosine-sulfated secreted form of eIF5A Oxidative Stress-Responsive Apoptosis-Inducing Protein (ORAIP). To investigate the role of ORAIP in a dextran sulfate sodium (DSS)-induced murine model of ulcerative colitis (UC), we analyzed the effects of in vivo treatment with anti-ORAIP neutralizing monoclonal antibody (mAb) on the DSS-induced disease exacerbation. The body weight in anti-ORAIP mAb-treated group was significantly heavier than that in a mouse IgG-treated control group on day 8 of DSS-treatment ((85.21 ± 1.03%) vs. (77.38 ± 2.07%); (mean ± SE0, n = 5 each, p < 0.01, t-test). In vivo anti-ORAIP mAb-treatment also significantly suppressed the shortening of colon length as well as Disease Activity Index (DAI) score ((5.00 ± 0.44) vs. (8.20 ± 0.37); (mean ± SE), n = 5 each, p < 0.001, t-test) by suppressing inflammation of the rectal tissue and apoptosis of intestinal mucosal cells. These data reveal the pivotal role of ORAIP in DSS-induced oxidative stress involved in an animal model of UC.


Sujet(s)
Rectocolite hémorragique , Sulfate dextran , Modèles animaux de maladie humaine , Stress oxydatif , Animaux , Sulfate dextran/toxicité , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Facteurs initiation chaîne peptidique/métabolisme , , Apoptose/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL
8.
BMC Cancer ; 24(1): 432, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38589831

RÉSUMÉ

BACKGROUND: One-third of diffuse large B-cell lymphoma (DLBCL) patients suffer relapse after standard treatment. Eukaryotic initiation factor 3a (eIF3a) is a key player in the initial stage of translation, which has been widely reported to be correlated with tumorigenesis and therapeutic response. This study aimed to explore the biological role of eIF3a, evaluate its prognostic and therapeutic potential in DLBCL. METHODS: RNA-seq datasets from GEO database were utilized to detect the expression and prognostic role of eIF3a in DLBCL patients. Protein level of eIF3a was estimated by western blot and immunohistochemical. Next, DLBCL cells were transfected with lentiviral vector either eIF3a-knockdown or empty to assess the biological role of eIF3a. Then, samples were divided into 2 clusters based on eIF3a expression and differentially expressed genes (DEGs) were identified. Function enrichment and mutation analysis of DEGs were employed to detect potential biological roles. Moreover, we also applied pan-cancer and chemosensitivity analysis for deep exploration. RESULTS: eIF3a expression was found to be higher in DLBCL than healthy controls, which was associated with worse prognosis. The expression of eIF3a protein was significantly increased in DLBCL cell lines compared with peripheral blood mononuclear cells (PBMCs) from healthy donors. eIF3a knockdown inhibited the proliferation of DLBCL cells and the expression of proliferation-related proteins and increase cell apoptosis rate. Besides, 114 DEGs were identified which had a close linkage to cell cycle and tumor immune. eIF3a and DEGs mutations were found to be correlated to chemosensitivity and vital signal pathways. Pan-cancer analysis demonstrated that high eIF3a expression was associated with worse prognosis in several tumors. Moreover, eIF3a expression was found to be related to chemosensitivity of several anti-tumor drugs in DLBCL, including Vincristine and Wee1 inhibitor. CONCLUSIONS: We firstly revealed the high expression and prognostic role of eIF3a in DLBCL, and eIF3a might promote the development of DLBCL through regulating cell proliferation and apoptosis. eIF3a expression was related to immune profile and chemosensitivity in DLBCL. These results suggest that eIF3a could serve as a potential prognostic biomarker and therapeutic target in DLBCL.


Sujet(s)
Antinéoplasiques , Lymphome B diffus à grandes cellules , Humains , Agranulocytes , Prolifération cellulaire/génétique , Antinéoplasiques/usage thérapeutique , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/diagnostic , Facteurs initiation chaîne peptidique/pharmacologie , Facteurs initiation chaîne peptidique/usage thérapeutique , Lignée cellulaire tumorale
9.
Wiley Interdiscip Rev RNA ; 15(2): e1833, 2024.
Article de Anglais | MEDLINE | ID: mdl-38433101

RÉSUMÉ

Selection of the correct start codon is critical for high-fidelity protein synthesis. In eukaryotes, this is typically governed by a multitude of initiation factors (eIFs), including eIF2·GTP that directly delivers the initiator tRNA (Met-tRNAi Met ) to the P site of the ribosome. However, numerous reports, some dating back to the early 1970s, have described other initiation factors having high affinity for the initiator tRNA and the ability of delivering it to the ribosome, which has provided a foundation for further work demonstrating non-canonical initiation mechanisms using alternative initiation factors. Here we provide a critical analysis of current understanding of eIF2A, eIF2D, and the MCT-1·DENR dimer, the evidence surrounding their ability to initiate translation, their implications in human disease, and lay out important key questions for the field. This article is categorized under: RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes Translation > Mechanisms Translation > Regulation.


Sujet(s)
Facteurs d'initiation eucaryotes , ARN de transfert de la méthionine , Ribosomes , Humains , Eucaryotes , Facteurs initiation chaîne peptidique , Ribosomes/génétique , ARN , Facteur-2 d'initiation eucaryote
10.
PLoS Pathog ; 20(2): e1011535, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38335237

RÉSUMÉ

A better mechanistic understanding of virus-host dependencies can help reveal vulnerabilities and identify opportunities for therapeutic intervention. Of particular interest are essential interactions that enable production of viral proteins, as those could target an early step in the virus lifecycle. Here, we use subcellular proteomics, ribosome profiling analyses and reporter assays to detect changes in protein synthesis dynamics during SARS-CoV-2 (CoV2) infection. We identify specific translation factors and molecular chaperones that are used by CoV2 to promote the synthesis and maturation of its own proteins. These can be targeted to inhibit infection, without major toxicity to the host. We also find that CoV2 non-structural protein 1 (Nsp1) cooperates with initiation factors EIF1 and 1A to selectively enhance translation of viral RNA. When EIF1/1A are depleted, more ribosomes initiate translation from a conserved upstream CUG start codon found in all genomic and subgenomic viral RNAs. This results in higher translation of an upstream open reading frame (uORF1) and lower translation of the main ORF, altering the stoichiometry of viral proteins and attenuating infection. Replacing the upstream CUG with AUG strongly inhibits translation of the main ORF independently of Nsp1, EIF1, or EIF1A. Taken together, our work describes multiple dependencies of CoV2 on host biosynthetic networks and proposes a model for dosage control of viral proteins through Nsp1-mediated control of translation start site selection.


Sujet(s)
COVID-19 , ARN viral , Humains , ARN viral/génétique , SARS-CoV-2/génétique , COVID-19/génétique , Facteurs initiation chaîne peptidique , Protéines virales
11.
Int J Mol Sci ; 25(3)2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38339043

RÉSUMÉ

Programmed ribosomal frameshifting (PRF) exists in all branches of life that regulate gene expression at the translational level. The eukaryotic translation initiation factor 5A (eIF5A) is a highly conserved protein essential in all eukaryotes. It is identified initially as an initiation factor and functions broadly in translation elongation and termination. The hypusination of eIF5A is specifically required for +1 PRF at the shifty site derived from the ornithine decarboxylase antizyme 1 (OAZ1) in Saccharomyces cerevisiae. However, whether the regulation of +1 PRF by yeast eIF5A is universal remains unknown. Here, we found that Sc-eIF5A depletion decreased the putrescine/spermidine ratio. The re-introduction of Sc-eIF5A in yeast eIF5A mutants recovered the putrescine/spermidine ratio. In addition, the Sc-eIF5A depletion decreases +1 PRF during the decoding of Ty1 retrotransposon mRNA, but has no effect on -1 PRF during the decoding of L-A virus mRNA. The re-introduction of Sc-eIF5A in yeast eIF5A mutants restored the +1 PRF rate of Ty1. The inhibition of the hypusine modification of yeast eIF5A by GC7 treatment or by mutating the hypusination site Lys to Arg caused decreases of +1 PRF rates in the Ty1 retrotransposon. Furthermore, mutational studies of the Ty1 frameshifting element support a model where the efficient removal of ribosomal subunits at the first Ty1 frame 0 stop codon is required for the frameshifting of trailing ribosomes. This dependency is likely due to the unique position of the frame 0 stop codon distance from the slippery sequence of Ty1. The results showed that eIF5A is a trans-regulator of +1 PRF for Ty1 retrotransposon and could function universally in yeast.


Sujet(s)
Décalage ribosomique , Saccharomyces cerevisiae , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Spermidine/métabolisme , Putrescine/métabolisme , Rétroéléments/génétique , Codon stop/génétique , Codon stop/métabolisme , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/métabolisme
12.
Microbiol Spectr ; 12(3): e0277323, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38329351

RÉSUMÉ

A significant increase of hospital-acquired bacterial infections during the COVID-19 pandemic has become an urgent medical problem. Clostridioides difficile is an urgent antibiotic-resistant bacterial pathogen and a leading causative agent of nosocomial infections. The increasing recurrence of C. difficile infection and antibiotic resistance in C. difficile has led to an unmet need for the discovery of new compounds distinctly different from present antimicrobials, while antimicrobial peptides as promising alternatives to conventional antibiotics have attracted growing interest recently. Protein synthesis is an essential metabolic process in all bacteria and a validated antibiotic target. Initiation factor 1 from C. difficile (Cd-IF1) is the smallest of the three initiation factors that acts to establish the 30S initiation complex to initiate translation during protein biosynthesis. Here, we report the solution nuclear magnetic resonance (NMR) structure of Cd-IF1 which adopts a typical ß-barrel fold and consists of a five-stranded ß-sheet and one short α-helix arranged in the sequential order ß1-ß2-ß3-α1-ß4-ß5. The interaction of Cd-IF1 with the 30S ribosomal subunit was studied by NMR titration for the construction of a structural model of Cd-IF1 binding with the 30S subunit. The short α-helix in IF1 was found to be critical for IF1 ribosomal binding. A peptide derived from this α-helix was tested and displayed a high ability to inhibit the growth of C. difficile and other bacterial strains. These results provide a clue for the rational design of new antimicrobials.IMPORTANCEBacterial infections continue to represent a major worldwide health hazard due to the emergence of drug-resistant strains. Clostridioides difficile is a common nosocomial pathogen and the causative agent in many infections resulting in an increase in morbidity and mortality. Bacterial protein synthesis is an essential metabolic process and an important target for antibiotic development; however, the precise structural mechanism underlying the process in C. difficile remains unknown. This study reports the solution structure of C. difficile translation initiation factor 1 (IF1) and its interaction with the 30S ribosomal subunit. A short α-helix in IF1 structure was identified as critically important for ribosomal binding and function in regulating the translation initiation, which allowed a rational design of a new peptide. The peptide demonstrated a high ability to inhibit bacterial growth with broad-spectrum antibacterial activity. This study provides a new clue for the rational design of new antimicrobials against bacterial infections.


Sujet(s)
Infections bactériennes , Clostridioides difficile , Humains , Peptides antimicrobiens , Cadmium , Pandémies , Facteurs initiation chaîne peptidique , Antibactériens/pharmacologie
13.
J Ethnopharmacol ; 325: 117859, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38316218

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Endometriosis (EMs) is characterized by inflammatory lesions, dysmenorrhea, infertility, and chronic pelvic pain. Single-target medications often fail to provide systemic therapeutic results owing to the complex mechanism underlying endometriosis. Although traditional Chinese medicines-such as Juan-Tong-Yin (JTY)-have shown promising results, their mechanisms of action remain largely unknown. AIM OF THE STUDY: To elucidate the therapeutic mechanism of JTY in EMs, focusing on endoplasmic reticulum (ER) stress-induced autophagy. MATERIALS AND METHODS: The major components of JTY were detected using high-performance liquid chromatography-mass spectrometry (HPLC-MS). The potential mechanism of JTY in EMs treatment was predicted using network pharmacological analysis. Finally, the pathogenesis of EMs was validated in a clinical case-control study and the molecular mechanism of JTY was validated in vitro using endometrial stromal cells (ESCs). RESULTS: In total, 241 compounds were analyzed and identified from JTY using UPLC-MS. Network pharmacology revealed 288 targets between the JTY components and EMs. Results of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses indicated that regulating autophagy, migration, apoptosis, and inflammation were the key mechanisms of JTY in treating EMs. Meanwhile, we found that protein kinase R-like endoplasmic reticulum kinase (PERK), Beclin-1, and microtubule-associated protein light chain 3 B (LC3B) expressions were lower in endometria of patients with EMs than in those with normal eutopic endometria (p < 0.05). Additionally, during in vitro experiments, treatment with 20% JTY-containing serum significantly suppressed ESC proliferation, achieving optimal effects after 48 h. Electron microscopy revealed significantly increased autophagy flux in the JTY group compared with the control group. Moreover, JTY treatment significantly reduced the migratory and invasive abilities of ESCs and upregulated protein expression of PERK, eukaryotic initiation factor 2α (eIF2α)/phospho-eukaryotic initiation factor 2α (p-eIF2α), activating Transcription Factor-4 (ATF4), Beclin-1, and LC3BII/I, while subsequently downregulating NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and interleukin 18 (IL-18) expression. However, administration of GSK2656157-a highly selective PERK inhibitor-reversed these changes. CONCLUSION: JTY ameliorates EMs by activating PERK associated with unfolded protein reaction, enhancing cell ER stress and autophagy, improving the inflammatory microenvironment, and decreasing the migration and invasion of ESCs.


Sujet(s)
Endométriose , Transduction du signal , Femelle , Humains , Bécline-1/métabolisme , Endométriose/anatomopathologie , Études cas-témoins , Chromatographie en phase liquide , Spectrométrie de masse en tandem , Stress du réticulum endoplasmique , Autophagie , Apoptose , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/pharmacologie
14.
Arch Biochem Biophys ; 754: 109921, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38341068

RÉSUMÉ

Zinc finger protein 131 (ZNF131), a member of BTB-ZF transcription factors, has been previously reported as an oncogene in several human cancers. However, the function and underlying mechanism of ZNF131 in hepatocellular carcinoma (HCC) are still unclear. In our study, the upregulated expression of ZNF131 mRNA was confirmed in HCC tissues by analyzing the TCGA and GEO datasets. The immunohistochemical staining data also revealed the overexpression of ZNF131 protein in HCC samples. High expression of ZNF131 predicted poor overall survival and disease-free survival in HCC patients. ZNF131 knockdown inhibited the proliferation and colony formation and led to G2/M phase arrest of HCC cells, while its overexpression promoted HCC cell proliferation, cell cycle progression and colony formation. Moreover, ZNF131 silencing repressed the growth of HCC cells in nude mice. Yes-associated protein 1 (YAP1) was recognized as an upstream regulator of ZNF131. Both YAP1 knockdown and inactivation reduced ZNF131 expression in HCC cells, and YAP1 overexpression enhanced ZNF131 level. Interestingly, we found that poly(A) binding protein interacting protein 1 (PAIP1) was a novel target of ZNF131. ZNF131 silencing downregulated while ZNF131 overexpression upregulated PAIP1 expression in HCC cells. The luciferase reporter assay demonstrated that ZNF131 regulated PAIP1 expression at the transcription level. Notably, we revealed that ZNF131 activated the AKT signaling by enhancing PAIP1 expression in HCC cells. AKT inhibitor markedly attenuated ZNF131-enhanced HCC cell proliferation. Restoring PAIP1 expression abrogated the inhibitory effects of ZNF131 knockdown on HCC cell proliferation and colony formation. To conclude, ZNF131 was highly expressed and acted as an oncogene in HCC. ZNF131, which was activated by YAP1, promoted HCC cell proliferation through transcriptional regulation of PAIP1.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Animaux , Souris , Humains , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Souris nude , Lignée cellulaire tumorale , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/métabolisme , Protéines de liaison à l'ARN/génétique
15.
Biol Reprod ; 110(4): 834-847, 2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38281153

RÉSUMÉ

Besides ubiquitous poly(A)-binding protein, cytoplasmic 1 (PABPC1), testis-specific PABPC2/PABPt (in humans, referred to as PABPC3), and female and male germline-specific PABPC1L/ePAB, have been reported in the mouse testis. Recent in silico analysis additionally identified testis-specific Pabpc6 in the mouse. In this study, we characterized PABPC6 and its mutant mice. PABPC6 was initially detectable in the cytoplasm of pachytene spermatocytes, increased in abundance in round spermatids, and decreased in elongating spermatids. PABPC6 was capable of binding to poly(A) tails of various mRNAs and interacting with translation-associated factors, including EIF4G, PAIP1, and PAIP2. Noteworthy was that PABPC6, unlike PABPC1, was barely associated with translationally active polysomes and enriched in chromatoid bodies of round spermatids. Despite these unique characteristics, neither synthesis of testicular proteins nor spermatogenesis was affected in the mutant mice lacking PABPC6, suggesting that PABPC6 is functionally redundant with other co-existing PABPC proteins during spermatogenesis.


Sujet(s)
Spermatogenèse , Testicule , Humains , Mâle , Souris , Femelle , Animaux , Testicule/métabolisme , Spermatogenèse/génétique , Spermatides/métabolisme , Protéines de liaison au poly(A)/génétique , Protéines de liaison au poly(A)/métabolisme , Cytoplasme/métabolisme , ARN messager/métabolisme , Facteurs initiation chaîne peptidique/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme
16.
Am J Respir Crit Care Med ; 209(11): 1376-1391, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38261723

RÉSUMÉ

Rationale: The ubiquitous polyamine spermidine is essential for cell survival and proliferation. One important function of spermidine is to serve as a substrate for hypusination, a posttranslational modification process that occurs exclusively on eukaryotic translation factor 5A (eIF5A) and ensures efficient translation of various gene products. Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive obliteration of the small pulmonary arteries (PAs) caused by excessive proliferation of PA smooth muscle cells (PASMCs) and suppressed apoptosis. Objectives: To characterize the role of hypusine signaling in PAH. Methods: Molecular, genetic, and pharmacological approaches were used both in vitro and in vivo to investigate the role of hypusine signaling in pulmonary vascular remodeling. Measurements and Main Results: Hypusine forming enzymes-deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH)-and hypusinated eukaryotic translation factor 5A are overexpressed in distal PAs and isolated PASMCs from PAH patients and animal models. In vitro, inhibition of DHPS using N1-guanyl-1,7-diaminoheptane or shRNA resulted in a decrease in PAH-PASMC resistance to apoptosis and proliferation. In vivo, inactivation of one allele of Dhps targeted to smooth muscle cells alleviates PAH in mice, and its pharmacological inhibition significantly decreases pulmonary vascular remodeling and improves hemodynamics and cardiac function in two rat models of established PAH. With mass spectrometry, hypusine signaling is shown to promote the expression of a broad array of proteins involved in oxidative phosphorylation, thus supporting the bioenergetic requirements of cell survival and proliferation. Conclusions: These findings support inhibiting hypusine signaling as a potential treatment for PAH.


Sujet(s)
Hypertension artérielle pulmonaire , Transduction du signal , Remodelage vasculaire , Animaux , Remodelage vasculaire/effets des médicaments et des substances chimiques , Remodelage vasculaire/physiologie , Rats , Humains , Hypertension artérielle pulmonaire/physiopathologie , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/métabolisme , Mâle , Modèles animaux de maladie humaine , Artère pulmonaire/physiopathologie , Artère pulmonaire/effets des médicaments et des substances chimiques , Souris , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/génétique , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , , Prolifération cellulaire/effets des médicaments et des substances chimiques , Oxidoreductases acting on CH-NH group donors/génétique , Oxidoreductases acting on CH-NH group donors/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/physiopathologie , Hypertension pulmonaire/physiopathologie , Hypertension pulmonaire/métabolisme , Lysine/analogues et dérivés
17.
EMBO Rep ; 25(3): 1415-1435, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38279019

RÉSUMÉ

Eukaryotic translation initiation factors have long been recognized for their critical roles in governing the translation of coding RNAs into peptides/proteins. However, whether they harbor functional activities at the post-translational level remains poorly understood. Here, we demonstrate that eIF3f1 (eukaryotic translation initiation factor 3 subunit f1), which encodes an archetypal deubiquitinase, is essential for the antimicrobial innate immune defense of Drosophila melanogaster. Our in vitro and in vivo evidence indicate that the immunological function of eIF3f1 is dependent on the N-terminal JAMM (JAB1/MPN/Mov34 metalloenzymes) domain. Mechanistically, eIF3f1 physically associates with dTak1 (Drosophila TGF-beta activating kinase 1), a key regulator of the IMD (immune deficiency) signaling pathway, and mediates the turnover of dTak1 by specifically restricting its K48-linked ubiquitination. Collectively, these results provide compelling insight into a noncanonical molecular function of a translation initiation factor that controls the post-translational modification of a target protein.


Sujet(s)
Protéines de Drosophila , Drosophila , Animaux , Drosophila/métabolisme , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Immunité innée , Facteurs initiation chaîne peptidique , Transduction du signal
18.
Biochimie ; 217: 31-41, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-36773835

RÉSUMÉ

In eukaryotes and in archaea late steps of translation initiation involve the two initiation factors e/aIF5B and e/aIF1A. These two factors are also orthologous to the bacterial IF2 and IF1 proteins, respectively. Recent cryo-EM studies showed how e/aIF5B and e/aIF1A cooperate on the small ribosomal subunit to favor the binding of the large ribosomal subunit and the formation of a ribosome competent for elongation. In this review, pioneering studies and recent biochemical and structural results providing new insights into the role of a/eIF5B in archaea and eukaryotes will be presented. Recent structures will also be compared to orthologous bacterial initiation complexes to highlight domain-specific features and the evolution of initiation mechanisms.


Sujet(s)
Facteur-1 d'initiation eucaryote , Facteurs initiation chaîne peptidique , Facteur-1 d'initiation eucaryote/analyse , Facteur-1 d'initiation eucaryote/composition chimique , Facteur-1 d'initiation eucaryote/métabolisme , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/analyse , Facteurs initiation chaîne peptidique/composition chimique , Bactéries/métabolisme , Ribosomes/métabolisme
19.
Histol Histopathol ; 39(4): 463-470, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37334930

RÉSUMÉ

BACKGROUND AND AIMS: Eukaryotic translation initiation factor 5A2 (EIF5A2) has been reported to be involved in metastasis and chemotherapy resistance in many human cancers. However, the effect and mechanism of EIF5A2 in oral cancer cells are unknown. Here, we investigated the effects of targeting EIF5A2 on chemotherapy resistance in oral cancer cells in vitro. METHODS: By using a lentiviral system, we investigated the effects of targeting EIF5A2 on the invasion, migration, growth, and chemosensitivity of SCC-9 cells to CDDP in vitro. Through the method of gene intervention, we explore the role of pro-apoptotic Bim and epithelial and mesenchymal marker E-cadherin protein in this process and the regulation of EIF5A2 on Bim and E-cadherin. RESULTS: Targeting EIF5A2 reduces invasion and migration in SCC-9 cells partly through upregulation of E-cadherin expression; Targeting EIF5A2 promotes cell apoptosis and inhibits cell survival as well as increasing chemosensitivity in SCC-9 cells through upregulation of Bim expression. CONCLUSION: EIF5A2 may be a novel potential therapeutic target for oral cancer by upregulation of Bim and E-cadherin.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs de la bouche , Humains , Facteurs initiation chaîne peptidique/métabolisme , Régulation positive , Cadhérines/génétique , Tumeurs de la bouche/traitement médicamenteux , Lignée cellulaire tumorale
20.
J Gastroenterol Hepatol ; 39(3): 596-607, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38059880

RÉSUMÉ

BACKGROUND AND AIM: Circular ubiquitin-like, containing PHD and ring finger domains 1 (circUHRF1) is aberrantly upregulated in human hepatocellular carcinoma (HCC) tissues. However, the underlying molecular mechanisms remain obscure. The present study aimed at elucidating the interactive function of circUHRF1-G9a-ubiquitin-like, containing PHD and ring finger domains 1 (UHRF1) mRNA-eukaryotic translation initiation factor 4A3 (EIF4A3)-PDZ and LIM domain 1 (PDLIM1) network in HCC. METHODS: Expression of circUHRF1, mRNAs of G9a, UHRF1, PDLIM1, epithelial-mesenchymal transition (EMT)-related proteins, and Hippo-Yap pathway components was determined by quantitative polymerase chain reaction (Q-PCR), immunofluorescence, or Western blot analysis. Tumorigenic and metastatic capacities of HCC cells were examined by cellular assays including Cell Counting Kit-8, colony formation, wound healing, and transwell assays. Molecular interactions between EIF4A3 and UHRF1 mRNA were detected by RNA pull-down experiment. Complex formation between UHRF1 and PDLIM1 promoter was detected by chromatin immunoprecipitation assay. Co-immunoprecipitation was performed to examine the binding between UHRF1 and G9a. RESULTS: Circular ubiquitin-like, containing PHD and ring finger domains 1, G9a, and UHRF1 were upregulated, while PDLIM1 was downregulated in HCC tissue samples and cell lines. Cellular silencing of circUHRF1 repressed HCC proliferation, invasion, migration, and EMT. G9a formed a complex with UHRF1 and inhibited PDLIM1 transcription. CONCLUSION: Eukaryotic translation initiation factor 4A3 regulated circUHRF1 expression by binding to UHRF1 mRNA promoter. circUHRF1 increased the stability of G9a and UHRF1 mRNAs through recruiting EIF4A3. Overexpression of circUHRF1 aggravated HCC progression through Hippo-Yap pathway and PDLIM1 inhibition. By elucidating the molecular function of circUHRF1-G9a-UHRF1 mRNA-EIF4A3-PDLIM1 network, our data shed light on the HCC pathogenesis and suggest a novel therapeutic strategy for future HCC treatment.


Sujet(s)
Carcinome hépatocellulaire , DEAD-box RNA helicases , Tumeurs du foie , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , ARN messager/génétique , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/usage thérapeutique , Ubiquitine/génétique , Ubiquitine/métabolisme , Ubiquitine/usage thérapeutique , Domaines à doigts de zinc de type RING , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/usage thérapeutique , Protéines liant les séquences stimulatrices de type CCAAT/composition chimique , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Facteurs initiation chaîne peptidique/génétique , Facteurs initiation chaîne peptidique/métabolisme , Facteurs initiation chaîne peptidique/usage thérapeutique , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/génétique , Facteur-4A d'initiation eucaryote/génétique , Facteur-4A d'initiation eucaryote/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...